Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Metab ; 22: 84-95, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30711402

RESUMEN

OBJECTIVE: Diabetes is a complex disease implicating several organs and cell types. Within the islets, dysregulation occurs in both alpha- and beta-cells, leading to defects of insulin secretion and increased glucagon secretion. Dysregulation of alpha-cells is associated with transcriptome changes. We hypothesized that microRNAs (miRNAs) which are negative regulators of mRNA stability and translation could be involved in alpha-cell alterations or adaptations during type 2 diabetes. METHODS: miRNA microarray analyses were performed on pure alpha- and beta-cells from high-fat diet fed obese hyperglycemic mice and low-fat diet fed controls. Then, the most regulated miRNA was overexpressed or inhibited in primary culture of mouse and human alpha-cells to determine its molecular and functional impact. RESULTS: 16 miRNAs were significantly regulated in alpha-cells of obese hyperglycemic mice and 28 in beta-cells. miR-132-3p had the strongest regulation level in alpha-cells, where it was downregulated, while we observed an opposite upregulation in beta-cells. In vitro experiments showed that miR-132-3p, which is inversely regulated by somatostatin and cAMP, is a positive modulator of alpha-cell proliferation and implicated in their resistance to apoptosis. These effects are associated with the regulation of a series of genes, including proliferation and stress markers Mki67 and Bbc3 in mouse and human alpha-cells, potentially involved in miR-132-3p functions. CONCLUSIONS: Downregulation of miR-132-3p in alpha-cells of obese diabetic mice may constitute a compensatory mechanism contributing to keep glucagon-producing cell number constant in diabetes.


Asunto(s)
Regulación hacia Abajo , Células Secretoras de Glucagón/metabolismo , MicroARNs/metabolismo , Obesidad/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Ratones Obesos
2.
Proc Natl Acad Sci U S A ; 113(33): E4847-56, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27482114

RESUMEN

Inflammation is characterized by the recruitment of leukocytes from the bloodstream. The rapid arrival of neutrophils is followed by a wave of inflammatory lymphocyte antigen 6 complex (Ly6C)-positive monocytes. In contrast Ly6C(low) monocytes survey the endothelium in the steady state, but their role in inflammation is still unclear. Here, using confocal intravital microscopy, we show that upon Toll-like receptor 7/8 (TLR7/8)-mediated inflammation of mesenteric veins, platelet activation drives the rapid mobilization of Ly6C(low) monocytes to the luminal side of the endothelium. After repeatedly interacting with platelets, Ly6C(low) monocytes commit to a meticulous patrolling of the endothelial wall and orchestrate the subsequent arrival and extravasation of neutrophils through the production of proinflammatory cytokines and chemokines. At a molecular level, we show that cysteine-rich protein 61 (CYR61)/CYR61 connective tissue growth factor nephroblastoma overexpressed 1 (CCN1) protein is released by activated platelets and enables the recruitment of Ly6C(low) monocytes upon vascular inflammation. In addition endothelium-bound CCN1 sustains the adequate patrolling of Ly6C(low) monocytes both in the steady state and under inflammatory conditions. Blocking CCN1 or platelets with specific antibodies impaired the early arrival of Ly6C(low) monocytes and abolished the recruitment of neutrophils. These results refine the leukocyte recruitment cascade model by introducing endothelium-bound CCN1 as an inflammation mediator and by demonstrating a role for platelets and patrolling Ly6C(low) monocytes in acute vascular inflammation.


Asunto(s)
Antígenos Ly/análisis , Proteína 61 Rica en Cisteína/fisiología , Monocitos/fisiología , Vasculitis/etiología , Animales , Plaquetas/fisiología , Movimiento Celular , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 8/fisiología
3.
Endocrinology ; 157(10): 3832-3843, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27547850

RESUMEN

Glucose homeostasis depends on the coordinated secretion of glucagon, insulin, and Glucagon-like peptide (GLP)-1 by pancreas and intestine. Obesity, which is associated with an increased risk of developing insulin resistance and type 2 diabetes, affects the function of these organs. Here, we investigate the functional and molecular adaptations of proglucagon-producing cells in obese mice to better define their involvement in type 2 diabetes development. We used GLU-Venus transgenic male mice specifically expressing Venus fluorochrome in proglucagon-producing cells. Mice were subjected to 16 weeks of low-fat diet or high-fat diet (HFD) and then subdivided by measuring glycated hemoglobin (HbA1c) in 3 groups: low-fat diet mice and I-HFD (glucose-intolerant) mice with similar HbA1c and H-HFD (hyperglycemic) mice, which exhibited higher HbA1c. At 16 weeks, both HFD groups exhibited similar weight gain, hyperinsulinemia, and insulin resistance. However, I-HFD mice exhibited better glucose tolerance compared with H-HFD mice. I-HFD mice displayed functional and molecular adaptations of enteroendocrine L-cells resulting in increased intestinal GLP-1 biosynthesis and release as well as maintained pancreatic α- and ß-cell functions. By contrast, H-HFD mice exhibited dysfunctional L, α- and ß-cells with increased ß- and L-cell numbers. Administration of the GLP-1R antagonist Exendin9-39 in I-HFD mice led to hyperglycemia and alterations of glucagon secretion without changes in insulin secretion. Our results highlight the cross-talk between islet and intestine endocrine cells and indicate that a compensatory adaptation of L-cell function in obesity plays an important role in preserving glucose homeostasis through the control of pancreatic α-cell functions.


Asunto(s)
Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Hiperglucemia/metabolismo , Obesidad/metabolismo , Animales , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Glucagón/metabolismo , Hiperglucemia/etiología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/complicaciones , Fragmentos de Péptidos , Fenotipo
4.
Endocrinology ; 157(2): 536-47, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26696123

RESUMEN

Glucagon and α-cell dysfunction are critical in the development of hyperglycemia during diabetes both in humans and rodents. We hypothesized that α-cell dysfunction leading to dysregulated glucagon secretion in diabetes is due to both a lack of insulin and intrinsic defects. To characterize α-cell dysfunction in diabetes, we used glucagon-Venus transgenic male mice and induced insulinopenic hyperglycemia by streptozotocin administration leading to alterations of glucagon secretion. We investigated the in vivo impact of insulinopenic hyperglycemia on glucagon-producing cells using FACS-sorted α-cells from control and diabetic mice. We demonstrate that increased glucagonemia in diabetic mice is mainly due to increases of glucagon release and biosynthesis per cell compared with controls without changes in α-cell mass. We identified genes coding for proteins involved in glucagon biosynthesis and secretion, α-cell differentiation, and potential stress markers such as the glucagon, Arx, MafB, cMaf, Brain4, Foxa1, Foxa3, HNF4α, TCF7L2, Glut1, Sglt2, Cav2.1, Cav2.2, Nav1.7, Kir6.2/Sur1, Pten, IR, NeuroD1, GPR40, and Sumo1 genes, which were abnormally regulated in diabetic mice. Importantly, insulin treatment partially corrected α-cell function and expression of genes coding for proglucagon, or involved in glucagon secretion, glucose transport and insulin signaling but not those coding for cMAF, FOXA1, and α-cell differentiation markers as well as GPR40, NEUROD1, CAV2.1, and SUMO1. Our results indicate that insulinopenic diabetes induce marked α-cell dysfunction and molecular alteration, which are only partially corrected by in vivo insulin treatment.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Insulina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diabetes Mellitus Experimental/patología , Glucagón/biosíntesis , Glucagón/genética , Insulina/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética
5.
J Immunol ; 194(2): 750-60, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25505285

RESUMEN

IL-33 is a cytokine of the IL-1 family, which signals through the ST2 receptor. Previous studies emphasized a role for IL-33 in shaping innate and adaptive immune responses. IL-33 was also reported to modulate myelopoiesis and myeloid cell activity. In this article, we describe IL-33-overexpressing CMV/IL33 and LysM/IL33 mice, which display an inflammatory phenotype associated with growth retardation and paw swelling. The phenotype of CMV/IL33 mice is dependent on activation of the ST2 receptor and is characterized by extensive neutrophil infiltration into different organs, including the paws. Local or systemic levels of proinflammatory mediators such as IL-1ß, Cxcl-1, G-CSF, and IL-6 are increased. CMV/IL-33 mice also suffer from anemia, thrombocytosis, and a marked dysregulation of myelopoiesis, leading to an important increase in myeloid cell production or accumulation in bone marrow (BM), spleen, and peripheral blood. Consistently, recombinant IL-33 induced proliferation of myeloid lineage cells in BM-derived granulocyte cultures, whereas IL-33 knockout mice exhibited minor deficiencies in spleen and BM myeloid cell populations. Our observations reveal a neutrophil-dominated inflammatory phenotype in IL-33-overexpressing CMV/IL33 and LysM/IL33 mice, and highlight important regulatory effects of IL-33 on myelopoiesis in vitro and in vivo, where excessive IL-33 signaling can translate into the occurrence of a myeloproliferative disorder.


Asunto(s)
Interleucinas/inmunología , Mielopoyesis/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Transducción de Señal/inmunología , Anemia/genética , Anemia/inmunología , Anemia/patología , Animales , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Expresión Génica , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-33 , Interleucina-6/genética , Interleucina-6/inmunología , Interleucinas/genética , Ratones , Ratones Noqueados , Mielopoyesis/genética , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología , Trastornos Mieloproliferativos/patología , Infiltración Neutrófila/genética , Neutrófilos/patología , Receptores de Interleucina/genética , Receptores de Interleucina/inmunología , Transducción de Señal/genética , Trombocitosis/genética , Trombocitosis/inmunología , Trombocitosis/patología
6.
Nat Commun ; 4: 2842, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24280864

RESUMEN

Thymic epithelial cells (TEC) are heterogeneous stromal cells that generate microenvironments required for the formation of T cells within the thymus. Defects in TEC lead to immunodeficiency or autoimmunity. Here we identify TEC as the major source of cysteine-rich protein 61 (CYR61), a matricellular protein implicated in cell proliferation and migration. Binding of CYR61 to LFA-1, ICAM-1 and integrin α6 supports the adhesion of TEC and thymocytes as well as their interaction. Treatment of thymic lobes with recombinant CYR61 expands the stromal compartment by inducing the proliferation of TEC and activates Akt signalling. Engraftment of CYR61-overexpressing thymic lobes into athymic nude mice drastically boosts the yield of thymic output via expansion of TEC. This increases the space for the recruitment of circulating hematopoietic progenitors and the development of T cells. Our discovery paves the way for therapeutic interventions designed to restore thymus stroma and T-cell generation.


Asunto(s)
Proteína 61 Rica en Cisteína/metabolismo , Células Epiteliales/citología , Células Madre/citología , Linfocitos T/citología , Timo/citología , Animales , Adhesión Celular , Proliferación Celular , Células Cultivadas , Proteína 61 Rica en Cisteína/genética , Células Epiteliales/metabolismo , Humanos , Integrina alfa6/genética , Integrina alfa6/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Unión Proteica , Células Madre/metabolismo , Linfocitos T/metabolismo , Timo/metabolismo
7.
Eur J Immunol ; 42(5): 1294-303, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22539301

RESUMEN

Interleukin-1 receptor antagonist (IL-1Ra) is a specific IL-1 inhibitor that possesses anti-inflammatory activities. Several studies in human and mouse suggested a protective role for IL-1Ra in liver inflammation, and we previously demonstrated that hepatocytes produce high levels of IL-1Ra in response to inflammatory challenge in vitro and in vivo. In the present study, we investigated the production and the biological function of hepatocyte-derived IL-1Ra in concanavalin A (ConA)-induced hepatitis in mice. We show that the injured liver produces large amounts of IL-1Ra and that secreted and intracellular IL-1Ra isoforms are produced with different kinetics during the course of hepatitis. By using hepatocyte-specific IL-1Ra-deficient mice (IL-1Ra(ΔH)), we demonstrate that hepatocytes represent the major cellular source of local IL-1Ra. Most interestingly, hepatic necrosis and inflammation were increased in IL-1Ra(ΔH) as compared with wild-type mice during the late phase of the disease, leading to a delayed resolution of hepatitis in IL-1Ra(ΔH) mice. In conclusion, our results show that the local production of IL-1Ra by hepatocytes contributes to the resolution of hepatitis.


Asunto(s)
Hepatitis/inmunología , Hepatocitos/inmunología , Proteína Antagonista del Receptor de Interleucina 1/inmunología , Animales , Concanavalina A/toxicidad , Citocinas/análisis , Hepatitis/genética , Hepatitis/patología , Hepatocitos/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/genética , Hígado/inmunología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis/inmunología
8.
J Immunol ; 177(5): 2899-907, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16920925

RESUMEN

Leptin-deficient ob/ob and leptin receptor (Ob-rb)-deficient db/db mice display a marked thymic atrophy and exhibit defective immune responses. Lymphocytes express leptin receptors and leptin exerts direct effects on T cells in vitro. In addition, ob/ob and db/db mice display multiple neuroendocrine and metabolic defects, through which leptin deficiency may indirectly affect the immune system in vivo. To study the relative contributions of direct and indirect effects of leptin on the immune system in a normal environment, we generated bone marrow chimeras (BMCs) by transplantation of leptin receptor-deficient db/db, or control db/+, bone marrow cells into wild-type (WT) recipients. The size and cellularity of the thymus, as well as cellular and humoral immune responses, were similar in db/db to WT and db/+ to WT BMCs. The immune phenotype of db/db mice is thus not explained by a cell autonomous defect of db/db lymphocytes. Conversely, thymus weight and cell number were decreased in the reverse graft setting in WT to db/db BMCs, indicating that expression of the leptin receptor in the environment is important for T cell development. Finally, normal thymocyte development occurred in fetal db/db thymi transplanted into WT hosts, indicating that direct effects of leptin are not required locally in the thymic microenvironment. In conclusion, direct effects of leptin on bone marrow-derived cells and on thymic stromal cells are not necessary for T lymphocyte maturation in normal mice. In contrast, leptin receptor deficiency affects the immune system indirectly via changes in the systemic environment.


Asunto(s)
Linfocitos/citología , Linfocitos/inmunología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Animales , Ganglios Linfáticos/citología , Recuento de Linfocitos , Masculino , Metilación , Ratones , Ratones Noqueados , Fenotipo , Receptores de Superficie Celular/genética , Receptores de Leptina , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/inmunología , Timo/citología , Timo/crecimiento & desarrollo , Timo/inmunología
9.
J Clin Invest ; 115(2): 237-46, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15650770

RESUMEN

Mechanisms regulating thrombus stabilization remain largely unknown. Here, we report that loss of any 1 of the Gas6 receptors (Gas6-Rs), i.e., Tyro3, Axl, or Mer, or delivery of a soluble extracellular domain of Axl that traps Gas6 protects mice against life-threatening thrombosis. Loss of a Gas6-R does not prevent initial platelet aggregation but impairs subsequent stabilization of platelet aggregates, at least in part by reducing "outside-in" signaling and platelet granule secretion. Gas6, through its receptors, activates PI3K and Akt and stimulates tyrosine phosphorylation of the beta3 integrin, thereby amplifying outside-in signaling via alphaIIbbeta3. Blocking the Gas6-R-alphaIIbbeta3 integrin cross-talk might be a novel approach to the reduction of thrombosis.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Agregación Plaquetaria/fisiología , Transducción de Señal/fisiología , Trombosis/metabolismo , Animales , Integrina beta3/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Agregación Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/genética , Trombosis/tratamiento farmacológico , Trombosis/genética , Trombosis/patología
10.
J Hepatol ; 41(5): 773-81, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15519650

RESUMEN

BACKGROUND/AIMS: Bacillus Calmette Guerin (BCG) infection causes hepatic injury following granuloma formation and secretion of cytokines which render mice highly sensitive to endotoxin-mediated hepatotoxicity. This work investigates the role of inducible nitric oxide synthase (iNOS) in liver damage induced by BCG and endotoxins in BCG-infected mice. METHODS: Liver injury and cytokine activation induced by BCG and by LPS upon BCG infection (BCG/LPS) were compared in wild-type and iNOS-/- mice. RESULTS: iNOS-/- mice infected with living BCG are protected from hepatic injury when compared to wild-type mice which express iNOS protein in macrophages forming hepatic granulomas. In addition, iNOS-/- mice show a decrease in BCG-induced IFN-gamma serum levels. LPS challenge in BCG-infected mice strongly activates iNOS in the liver and spleen of wild-type mice which show important liver damage associated with a dramatic increase in TNF and IL-6 and also Th1 type cytokines. In contrast, iNOS-/- mice are protected from liver injury after BCG/LPS challenge and their TNF, IL-6 and Th1 type cytokine serum levels raise moderately. CONCLUSIONS: These results demonstrate that nitric oxide (NO) from iNOS is involved in hepatotoxicity induced by both mycobacterial infection and endotoxin effects upon BCG infection and that inhibition of NO from iNOS protects from liver injuries.


Asunto(s)
Lipopolisacáridos/farmacología , Hepatopatías/metabolismo , Mycobacterium bovis , Óxido Nítrico Sintasa/genética , Tuberculosis/metabolismo , Animales , Interleucina-6/metabolismo , Hígado/enzimología , Hepatopatías/inmunología , Hepatopatías/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Bazo/enzimología , Células TH1/metabolismo , Tuberculosis/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 284(1): L197-204, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12388343

RESUMEN

We previously reported that exposure of mice to hyperoxia is characterized by extensive lung cell necrosis and apoptosis, mild inflammatory response, and elevated circulating levels of corticosterone. Administration of hydroxycortisone acetate during hyperoxia aggravated lung injury. Using adrenalectomized (ADX) and sham-operated (sham) mice, we studied the role of the glucocorticoids in hyperoxia-induced lung injury. Lung damage was attenuated in ADX mice as measured by lung weight and protein and cell content in bronchoalveolar lavage and as seen by light microscopy. Mortality was delayed by 10 h. Nuclear factor-kappaB (NF-kappaB) activity was significantly decreased in lungs of sham mice exposed to hyperoxia but was preserved in ADX mice. There was a correlation between NF-kappaB activity in ADX mice and decreased levels of IkappaBalpha. In contrast, activator protein-1 activity increased similarly in both groups of mice. Levels of interleukin-6 (IL-6), a transcriptional target of NF-kappaB, were higher in bronchoalveolar lavage and serum of ADX than sham mice. However, the protective effect of ADX was not mediated by IL-6, because administration of recombinant human IL-6 to sham mice did not prevent lung damage. These results demonstrate that the adrenal response aggravates alveolar injury and is likely to be mediated by the decrease of NF-kappaB function involved in cell survival.


Asunto(s)
Glucocorticoides/farmacología , Hiperoxia/metabolismo , Hiperoxia/patología , Pulmón/metabolismo , Pulmón/patología , FN-kappa B/antagonistas & inhibidores , Prolina/análogos & derivados , Adrenalectomía , Animales , Apoptosis , Corticosterona/metabolismo , Proteínas I-kappa B/metabolismo , Interleucina-6/farmacología , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Prolina/farmacología , Alveolos Pulmonares , Edema Pulmonar/prevención & control , Tiocarbamatos/farmacología , Timo/patología , Timo/fisiopatología , Factor de Transcripción AP-1/metabolismo
12.
Lab Invest ; 82(9): 1155-66, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12218076

RESUMEN

Infection of susceptible mice with Plasmodium berghei Anka leads to a syndrome of severe or cerebral malaria. Tumor necrosis factor (TNF) contributes to this syndrome, apparently by acting on its receptor 2 (TNFR2) because TNFR1-/- are susceptible, whereas TNFR2-/- mice are resistant. In this work, we confirmed the essential role of the TNFR2 in cerebral malaria because 6 to 8 days after Plasmodium berghei Anka infection, hypothermia, coma, and death were observed in +/+ or TNFR1-/-, but never in TNFR2-/-, mice. TNF production, evaluated by the serum levels or the mRNA levels in the brain, spleen or lung, was similar in +/+, TNFR1-/-, or TNFR2-/- mice. Macrophage or parasitized red blood cell sequestration in brain or lung was similar in TNFR1-/- and TNFR2-/- mice. Accordingly, up-regulation of CD54 or CD40 in brain or lung was also similar in TNFR1-/- or TNFR2-/- mice. Platelet loss, manifested by thrombocytopenia and the presence of microparticles in plasma, was similar in TNFR1-/- or TNFR2-/- mice. Breakdown of the blood-brain barrier, detected by the diffusion of tracers, was attenuated in both TNFR1-/- and TNFR2-/-, compared with +/+, mice. Endothelial cells from brain capillaries, examined by transmission electron microscopy, were similar in infected TNFR1-/- or TNFR2-/- mice, whereas the basement membrane was enlarged in TNFR1-/- mice. Hypothermic mice were also hyperglycemic, and this was evident in +/+ and TNFR1-/-, but not in TNFR2-/-, mice. In addition, infected +/+ and TNFR1-/- mice became insulin resistant, while in contrast TNFR2-/- became extremely insulin sensitive. This study supports the possibility that coma and death are mediated not by cell sequestration or breakdown of vascular permeability, similar in TNFR1-/- or TNFR2-/- mice, but by metabolic disturbances selectively mediated by the TNFR2.


Asunto(s)
Antígenos CD/fisiología , Malaria Cerebral/etiología , Receptores del Factor de Necrosis Tumoral/fisiología , Animales , Encéfalo/irrigación sanguínea , Antígenos CD4/genética , Antígenos CD40/genética , Ligando de CD40/genética , Permeabilidad Capilar , Coma/etiología , Eritrocitos/fisiología , Insulina/farmacología , Macrófagos/fisiología , Malaria Cerebral/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , ARN Mensajero/análisis , Receptores Tipo I de Factores de Necrosis Tumoral , Receptores Tipo II del Factor de Necrosis Tumoral , Trombocitopenia/etiología
13.
Eur J Haematol ; 68(5): 253-61, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12144531

RESUMEN

Anti-platelet antibodies are known to contribute to some types of thrombocytopenia. In this work we investigated anti-platelet antibodies with opposite influence upon activation and kinetics of platelet caspases. A rabbit anti-platelet antibody induced a profound thrombocytopenia, which was associated with an increase of microparticles in plasma and an activation of platelet caspases, as detected by the binding of a carboxyfluorescein-labeled fluoromethyl ketone probe (FAM-VAD-fmk). Furthermore, microparticles and thrombocytopenia were prevented by the injection of a caspase inhibitor ZVAD-fmk. In contrast, an anti-CD18 mAb (M18.2) induced a thrombocytosis, due to an increased platelet life-span and which was evident in wild-type (+/+), but not in CD18-/- or CD87-/-, mice indicating a requirement of these two surface molecules. Activation of caspases was decreased in platelets from mice injected with the M 18.2 mAb, as evidenced by a decreased binding of the VAD probe, detected by flow cytometry, or an increase in the level of pro-caspase-3, seen on Western blots. These observations indicate firstly, that anti-platelet antibodies can either promote or inhibit activation of platelet caspases, and secondly, that the activation of caspases regulates platelet life-span.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Plaquetas/inmunología , Antígenos CD18/inmunología , Caspasas/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/inmunología , Animales , Plaquetas/citología , Plaquetas/enzimología , Antígenos CD18/genética , Antígenos CD18/fisiología , Caspasas/metabolismo , Activación Enzimática/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/fisiología , Ratones , Ratones Noqueados , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Trombocitopenia/inducido químicamente , Trombocitopenia/enzimología , Trombocitopenia/inmunología
14.
Cytokine ; 18(4): 222-30, 2002 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-12126645

RESUMEN

TNF is known to induce a thrombocytopenia, due to a reduced platelet life span. Injection of TNF (10 microg) to mice did markedly increase the number of platelet-derived microparticles in plasma, most pronounced 1h after injection. Injection of TNF induced a transient activation of platelet caspases, -1, -3, -6, -8, -9, as seen by the binding of caspases probes detected by flow cytometry, most pronounced 1h after injection. Activation of caspase-3 was also evidenced by antibodies. Injection of the caspases inhibitor ZVAD-fmk decreased TNF-induced generation of microparticles and thrombocytopenia, indicating a causal role of caspases in platelet fragmentation. Activation of platelet caspases was also evident in platelets exposed to TNF in vitro, indicating that TNF acts on platelets directly. Comparison of platelets from +/+, TNFR1 -/- and TNFR2 -/- mice showed that caspases are activated mainly by the TNFR1. These observations indicate that TNF activates platelet caspases via the TNFR1, which results in platelet fragmentation and thrombocytopenia.


Asunto(s)
Plaquetas/enzimología , Caspasas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Animales , Proteínas del Linfoma 3 de Células B , Western Blotting , Caspasa 3 , Caspasas/sangre , Separación Celular , Activación Enzimática , Citometría de Flujo , Colorantes Fluorescentes/farmacología , Cinética , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Péptidos/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Recombinantes/metabolismo , Trombocitopenia/metabolismo , Factores de Tiempo , Factores de Transcripción
15.
Am J Pathol ; 160(1): 67-71, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11786400

RESUMEN

Recent studies suggest that apoptosis plays a role in oxygen-induced injury, although the activation pathways and the executioner proteases that lead to cleavage of lung cell proteins and DNA, are not yet identified. We explored previously the tumor necrosis factor/tumor necrosis factor receptor and the Fas/FasL, belonging to the intrinsic pathway, and could not demonstrate any protective effect by interfering with these cell receptors. Lately, it has been shown that interacting with the CD40 system, also known to promote cell death, by administering anti-CD40 ligand (L) antibody was beneficial in several diseases and, in particular, in hyperoxia-induced injury. Using CD40- and CD40L-deficient mice (-/-) as well as administering anti-CD40L antibody, we examined the extent of lung injury in oxygen-breathing mice by several ways (lung weight, histology, inflammatory mediators, and DNA ladder) as well as the mortality. The development of lung injury was similar in wild-type, CD40-/-, CD40L-/-, or in wild-type mice treated with anti-CD40L antibody. Apoptosis was present in all conditions at 72 hours of oxygen exposure. These results show that oxygen-induced injury does not require CD40-CD40L interaction and that apoptosis of lung cells does not involve this pathway.


Asunto(s)
Antígenos CD40/fisiología , Ligando de CD40/fisiología , Hiperoxia/patología , Pulmón/patología , Animales , Anticuerpos/farmacología , Apoptosis , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Antígenos CD40/genética , Ligando de CD40/genética , Ligando de CD40/inmunología , Citocinas/análisis , Hiperoxia/metabolismo , Hiperoxia/fisiopatología , Hiperoxia/prevención & control , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/genética , ARN Mensajero/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...